Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
J Minim Invasive Surg ; 26(1): 21-27, 2023 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-36936042

RESUMEN

Purpose: Esophagojejunostomy leakage after total gastrectomy for gastric cancer is one of the most serious and sometimes life-threatening adverse events. The purpose of this study was to evaluate complications after total gastrectomy in patients with gastric cancer during the period when Histoacryl (B. Braun) injection was performed. Therapeutic outcome of endoscopic Histoacryl injection for esophagojejunostomy leakage was also determined. Methods: This was a single-center retrospective study. Between January 2016 and December 2021, clinicopathologic characteristics and surgical outcomes of 205 patients who underwent total gastrectomy were investigated. Baseline characteristics and clinical outcomes of 10 patients with esophagojejunostomy leakage were also investigated. Results: Postoperative complication and mortality rates of total gastrectomy in 205 patients were 25.4% and 0.9%, respectively. Serious complications more than Clavien-Dindo IIIb accounted for 6.3%. Ten (4.9%) esophagojejunostomy leakages occurred in 205 patients. Among 10 esophagojejunostomy leakage patients, endoscopic Histoacryl injection was performed on eight patients and leakage was successfully managed with endoscopic Histoacryl injection in seven patients (87.5%). Mean postinjection hospital stay of seven successfully managed patients was 13.8 days. They were able to drink water at 1-6 days after injection. Among eight patients with endoscopic Histoacryl injection, six patients were injected once and two patients were injected three times. Conclusion: Endoscopic Histoacryl injection for esophagojejunostomy leakage after total gastrectomy can be considered as a useful treatment for some selected cases.

2.
Toxicol Res ; 38(4): 591-600, 2022 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-36277372

RESUMEN

2,4,3',5'-Tetramethoxystilbene (TMS) is a selective inhibitor of cytochrome P450 1B1 to block the conversion from estradiol to 4-OH-estradiol. Several studies suggested that TMS may act as a potent anti-cancer agent for hormone-related cancer including cervical cancer. Nutlin-3a is a cis-imidazoline analog that interferes with the interaction between mouse double minute 2 homolog (MDM2) and the tumor suppressor p53. The purpose of the study was to compare the cytotoxic effect of TMS and nutlin-3a treatment individually and in combination in HeLa cells. To assess the potential synergistic effects between TMS and nutlin-3a, low concentrations of TMS and nutlin-3a were simultaneously treated in HeLa cells. Based on cell viability, apoptosis assays, and the increase in cleaved caspase-3 and poly (ADP-ribose) polymerase cleavage, it was demonstrated that the combination with TMS and nutlin-3a exerts a synergistic effect on cancer cell death. Isobologram analysis of HeLa cells noted synergism between TMS and nutlin-3a. The combined treatment increased the expression of mitochondrial pro-apoptotic factors such as Bax and Bak, and decreased the expression of the XIAP. In addition, combination treatment significantly enhanced the translocation of AIF to the nucleus in HeLa cells. In conclusion, the results demonstrate that the combination of TMS and nutlin-3a induces synergistic apoptosis in HeLa cells, suggesting the possibility that this combination can be applied as a novel therapeutic strategy for cervical cancer.

3.
Sci Rep ; 11(1): 20867, 2021 10 21.
Artículo en Inglés | MEDLINE | ID: mdl-34675221

RESUMEN

Human steroid sulfatase (STS) is an enzyme that catalyzes the hydrolysis of dehydroepiandrosterone sulfate (DHEAS), estrone sulfate (E1S), and cholesterol sulfate. Abnormal expression of STS causes several diseases including colorectal, breast, and prostate cancer and refractory skin disease. In particular, accumulation of intracellular cholesterol sulfate by STS deficiency leads to a skin disorder with abnormal keratinization called X-linked ichthyosis (XLI). To determine the detailed mechanisms of XLI, we performed RNA sequencing (RNA-seq) analysis using human keratinocyte HaCaT cells treated with cholesterol and cholesterol sulfate. Of the genes with expression changes greater than 1.5-fold, Yippee-like 3 (YPEL3), a factor expected to affect cell differentiation, was found. Induction of YPEL3 causes permanent growth arrest, cellular senescence, and inhibition of metastasis in normal and tumor cells. In this study, we demonstrate that YPEL3 expression was induced by STS deficiency and, using the CRISPR/Cas9 system, a partial knock-out (STS+/-) cell line was constructed to establish a disease model for XLI studies. Furthermore, we show that increased expression of YPEL3 in STS-deficient cell lines promoted cellular senescence and expression of keratinization-related proteins such as involucrin and loricrin. Our results suggest that upregulation of YPEL3 expression by STS deficiency may play a crucial role in inducing cellular senescence and abnormal differentiation in human keratinocytes.


Asunto(s)
Ictiosis Ligada al Cromosoma X/genética , Queratinocitos/patología , Esteril-Sulfatasa/genética , Proteínas Supresoras de Tumor/genética , Sistemas CRISPR-Cas , Diferenciación Celular , Línea Celular , Senescencia Celular , Humanos , Ictiosis Ligada al Cromosoma X/patología , Queratinocitos/metabolismo , Regulación hacia Arriba
4.
Arch Pharm Res ; 44(1): 63-83, 2021 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-33484438

RESUMEN

Human cytochrome P450 enzymes (CYPs) play a critical role in various biological processes and human diseases. CYP1 family members, including CYP1A1, CYP1A2, and CYP1B1, are induced by aryl hydrocarbon receptors (AhRs). The binding of ligands such as polycyclic aromatic hydrocarbons activates the AhRs, which are involved in the metabolism (including oxidation) of various endogenous or exogenous substrates. The ligands that induce CYP1 expression are reported to be carcinogenic xenobiotics. Hence, CYP1 enzymes are correlated with the pathogenesis of cancers. Various endogenous substrates are involved in the metabolism of steroid hormones, eicosanoids, and other biological molecules that mediate the pathogenesis of several human diseases. Additionally, CYP1s metabolize and activate/inactivate therapeutic drugs, especially, anti-cancer agents. As the metabolism of drugs determines their therapeutic efficacy, CYP1s can determine the susceptibility of patients to some drugs. Thus, understanding the role of CYP1s in diseases and establishing novel and efficient therapeutic strategies based on CYP1s have piqued the interest of the scientific community.


Asunto(s)
Citocromo P-450 CYP1A1/metabolismo , Citocromo P-450 CYP1A2/metabolismo , Citocromo P-450 CYP1B1/metabolismo , Xenobióticos/farmacocinética , Animales , Citocromo P-450 CYP1A1/genética , Citocromo P-450 CYP1A2/genética , Citocromo P-450 CYP1B1/genética , Modelos Animales de Enfermedad , Predisposición Genética a la Enfermedad , Humanos , Ligandos , Receptores de Hidrocarburo de Aril/metabolismo
5.
J Toxicol Environ Health A ; 84(2): 84-94, 2021 01 17.
Artículo en Inglés | MEDLINE | ID: mdl-33103613

RESUMEN

Auranofin is a gold complex used as an anti-rheumatic agent and may act as a potent anticancer drug against breast tumors. Trametinib is a specific mitogen-activated protein kinase inhibitor, approved for the treatment of metastatic melanoma. The aim of this study was to examine the synergistic effects of auranofin and trametinib on apoptosis in MCF-7 human breast cancer cells. The combination treatment inhibited cancer cell proliferation and induced cell cycle arrest at the sub-G1 phase and apoptosis via poly (ADP-ribose) polymerase cleavage and caspase-3/7 activation. It is noteworthy that this treatment significantly increased p38 mitogen-activated protein kinase (MAPK) phosphorylation to induce mitochondrial stress, subsequently promoting cancer cell apoptosis through release of apoptosis-inducing factor. Further data demonstrated that combined treatment significantly induced increase in nuclear translocation of AIF. These results indicated that activation of the p38 MAPK signaling pathway and mitochondrial apoptosis may contribute to the synergistic consequences in MCF-7 cells. Collectively, our data demonstrated that combined treatment with auranofin and trametinib exhibited synergistic breast cancer cell death and this combination might be utilized as a novel therapeutic strategy for breast cancer.


Asunto(s)
Antineoplásicos/farmacología , Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Auranofina/farmacología , Neoplasias de la Mama/tratamiento farmacológico , Piridonas/farmacología , Pirimidinonas/farmacología , Apoptosis/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Femenino , Humanos , Células MCF-7 , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
6.
Biomol Ther (Seoul) ; 28(5): 473-481, 2020 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-32536618

RESUMEN

Axl receptor tyrosine kinase has been implicated in cancer progression, invasion, and metastasis in various cancer types. Axl overexpression has been observed in many cancers, and selective inhibitors of Axl, including R428, may be promising therapeutic agents for several human cancers, such as breast, lung, and pancreatic cancers. Here, we examined the cell growth inhibition mediated by R428 and auranofin individually as well as in combination in the human breast cancer cell lines MCF-7 and MDAMB- 231 to identify new advanced combination treatments for human breast cancer. Our data showed that combination therapy with R428 and auranofin markedly inhibited cancer cell proliferation. Isobologram analyses of these cells indicated a clear synergism between R428 and auranofin with a combination index value of 0.73. The combination treatment promoted apoptosis as indicated by caspase 3 activation and poly (ADP-ribose) polymerase cleavage. Cancer cell migration was also significantly inhibited by this combination treatment. Moreover, we found that combination therapy significantly increased the expression level of Bax, a mitochondrial proapoptotic factor, but decreased that of the X-linked inhibitor of apoptosis protein. Furthermore, the suppression of cell viability and induction of Bax expression by the combination treatment were recovered by treatment with N-acetylcysteine. In conclusion, our data demonstrated that combined treatment with R428 and auranofin synergistically induced apoptosis in human breast cancer cells and may thus serve as a novel and valuable approach for cancer therapy.

7.
Biochim Biophys Acta Mol Basis Dis ; 1865(9): 2464-2474, 2019 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-31195119

RESUMEN

Human steroid sulfatase (STS) has been linked with poor prognosis in steroid-associated tumors and represents an important clinical target in cancers, yet the mechanism of STS-induced carcinogenesis remains unclear. To correlate STS with cancer metabolism, we determined the effects of STS on aerobic glycolysis. STS overexpression increased cellular levels of lactic acid, the final product of aerobic glycolysis. Moreover, STS suppressed the oxygen consumption rate (OCR), which represents mitochondrial respiration. Inhibition of STS by the specific inhibitor STX064 recovered STS-induced OCR repression and lactic acid over-production. DHEA, but not DHEA-S, suppressed the OCR level and enhanced lactic acid production. To understand the molecular mechanism of STS-induced cancer metabolism, we measured the expression of glycolytic enzymes hexokinase 2 (HK2) and pyruvate kinase M2 (PKM2), which was highly upregulated by STS and DHEA at both protein and mRNA levels. HIF1α is a key mediator of aerobic glycolysis, and STS enhanced HIF1α promoter activity, mRNA expression, and protein expression. Down-regulation of HIF1α by siRNA suppressed the HK2 and PKM2 expression induced by both STS and DHEA. HIF1α siRNA also recovered the OCR repression and lactic acid over-production induced by both STS and DHEA. To explore the mechanism in vivo, we produced transgenic mice overexpressing STS and found that STS expression was particularly enhanced in the lung. Consistent with our in vitro results, the expression of HIF1α, HK2, and PKM2 was also increased in mouse lung tissues. In conclusion, we suggest that STS may induce aerobic glycolysis through enhancing HIF1α expression.


Asunto(s)
Proteínas Portadoras/metabolismo , Glucólisis , Hexoquinasa/metabolismo , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Proteínas de la Membrana/metabolismo , Esteril-Sulfatasa/metabolismo , Hormonas Tiroideas/metabolismo , Animales , Proteínas Portadoras/genética , Deshidroepiandrosterona/farmacología , Inhibidores Enzimáticos/farmacología , Femenino , Glucólisis/efectos de los fármacos , Células HeLa , Hexoquinasa/genética , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/antagonistas & inhibidores , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Pulmón/metabolismo , Proteínas de la Membrana/genética , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Consumo de Oxígeno/efectos de los fármacos , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , Esteril-Sulfatasa/antagonistas & inhibidores , Hormonas Tiroideas/genética , Proteínas de Unión a Hormona Tiroide
8.
J Vet Med Sci ; 81(2): 160-164, 2019 Feb 09.
Artículo en Inglés | MEDLINE | ID: mdl-30568051

RESUMEN

This study examined and compared the branching pattern of the aortic arch (AA) and its major branches in the Siberian roe deer (Capreolus pygargus) on Jeju Island (Jeju roe deer [JRD]) with those in the roe deer of the Korean peninsula (mainland roe deer [MRD]). Seven of the nine expected types was observed in the arterial silicone casts of 29 deer (10 males, 19 females). The JRD was identical to the MRD in that absence of the typical pattern; however, the main three pattern types differed between the two. This difference resulted from differences in the branching patterns of the right subclavian artery and costocervical trunk. In conclusion, the JRD has different type of AA from the MRD.


Asunto(s)
Aorta Torácica/anatomía & histología , Ciervos/anatomía & histología , Animales , Tronco Braquiocefálico/anatomía & histología , Femenino , Masculino , República de Corea , Arteria Subclavia/anatomía & histología
9.
Toxicol Sci ; 165(2): 335-346, 2018 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-29893927

RESUMEN

Numerous studies have attempted to develop a new in vitro eye irritation test (EIT). To obtain more reliable results from EIT, potential new biomarkers that reflect eye irritation by chemicals must be identified. We investigated candidate biomarkers for eye irritation, using a proteomics approach. Sodium lauryl sulfate (SLS) or benzalkonium chloride (BAC) was applied on a reconstructed human cornea-like epithelium model, MCTT HCE, and corneal protein expression was examined by two-dimensional gel electrophoresis. We found that ezrin (EZR) was significantly upregulated by SLS or BAC. In addition, upregulation of EZR in immortalized human corneal cells treated with SLS or BAC was confirmed by quantitative reverse transcription-PCR and western blot analysis. Furthermore, other well-known eye irritants such as cetylpyridinium bromide, Triton X-100, cyclohexanol, ethanol, 2-methyl-1-pentanol, and sodium hydroxide significantly increased EZR expression in immortalized human corneal cells. Induction of EZR promoter activity in irritant-treated human corneal cells was confirmed by a luciferase gene reporter assay. In conclusion, EZR expression may be a potential biomarker for detecting eye irritation, which may substantially improve the performance of in vitro EIT.


Asunto(s)
Proteínas del Citoesqueleto/biosíntesis , Células Epiteliales/efectos de los fármacos , Epitelio Corneal/efectos de los fármacos , Irritantes/toxicidad , Modelos Biológicos , Pruebas de Toxicidad/métodos , Compuestos de Benzalconio/farmacología , Biomarcadores/metabolismo , Técnicas de Cultivo de Célula , Supervivencia Celular/efectos de los fármacos , Proteínas del Citoesqueleto/genética , Células Epiteliales/metabolismo , Epitelio Corneal/metabolismo , Expresión Génica/efectos de los fármacos , Humanos , Dodecil Sulfato de Sodio/farmacología
10.
J Vet Med Sci ; 80(1): 128-132, 2018 Feb 02.
Artículo en Inglés | MEDLINE | ID: mdl-29199212

RESUMEN

This study examined the branching pattern of the aortic arch (AA) and its major branches in the Siberian roe deer (Capreolus pygargus Pallas, 1771) from South Korea. A total of eight of the nine expected types, based on the branching site and bilateral levels of the costocervical trunk (CCT) and subclavian artery (SB), were observed in the arterial silicone casts of 35 deer (16 males, 19 females). This deer has no typical type. The three most common types were present in 28.6, 25.7 and 20.0% of cases and resulted from different branching patterns of the left CCT and left SB. These results suggest that the Siberian roe deer in the Korean peninsula has various AA branching patterns, which differs from other ruminants.


Asunto(s)
Aorta Torácica/anatomía & histología , Ciervos/anatomía & histología , Animales , Arteria Carótida Común/anatomía & histología , Femenino , Masculino , Arterias Mamarias/anatomía & histología , República de Corea , Arteria Subclavia/anatomía & histología
11.
Oncotarget ; 8(43): 74263-74275, 2017 Sep 26.
Artículo en Inglés | MEDLINE | ID: mdl-29088783

RESUMEN

Annexin A5 (ANXA5) is a member of the annexin protein family. Previous studies have shown that ANXA5 is involved in anti-inflammation and cell death. However, the detailed mechanism of the role of ANXA5 in cancer cells is not well understood. In this study, we investigated the inhibitory effect of ANXA5 on cyclooxygenase-2 (COX-2) in prostate cancer cells. Expression of COX-2 induced by TNF-α was inhibited by overexpression of ANXA5 and inhibition of COX-2 expression by auranofin, which could induce ANXA5 expression, was restored by ANXA5 knockdown. In addition, ANXA5 knockdown induces phosphorylation of NF-κB p65 in prostate cancer cells, indicating that ANXA5 causes COX-2 downregulation through inhibition of p65 activation. We also found that protein kinase C (PKC)-ζ protein levels were upregulated by the inhibition of ANXA5, although the mRNA levels were unaffected. We have shown that upregulated COX-2 expression by inhibition of ANXA5 is attenuated by PKC-ζ siRNA. In summary, this study demonstrates that downregulation of PKC-ζ-NF-κB signaling by ANXA5 may inhibit COX-2 expression in prostate cancer.

12.
Oncotarget ; 8(37): 61604-61617, 2017 Sep 22.
Artículo en Inglés | MEDLINE | ID: mdl-28977889

RESUMEN

Steroid sulfatase (STS) catalyzes the hydrolysis of estrone sulfate and dehydroepiandrosterone sulfate (DHEAS) to their unconjugated biologically active forms. Although STS is considered a therapeutic target for estrogen-dependent diseases, the cellular functions of STS remain unclear. We found that STS induces Wnt/ß-catenin s Delete ignaling in PC-3 and HeLa cells. STS increases levels of ß-catenin, phospho-ß-catenin, and phospho-GSK3ß. Enhanced translocation of ß-catenin to the nucleus by STS might activate transcription of target genes such as cyclin D1, c-myc, and MMP-7. STS knockdown by siRNA resulted in downregulation of Wnt/ß-catenin signaling. ß-Catenin/TCF-mediated transcription was also enhanced by STS. STS induced an epithelial-mesenchymal transition (EMT) as it reduced the levels of E-cadherin, whereas levels of mesenchymal markers such as N-cadherin and vimentin were enhanced. We found that STS induced Twist1 expression through HIFα activation as HIF-1α knockdown significantly blocks the ability of STS to induce Twist1 transcription. Furthermore, DHEA, but not DHEAS is capable of inducing Twist1. Treatment with a STS inhibitor prevented STS-mediated Wnt/ß-catenin signaling and Twist1 expression. Interestingly, cancer cell migration, invasion, and MMPs expression induced by STS were also inhibited by a STS inhibitor. Taken together, these results suggest that STS induces Wnt/ß-catenin signaling and EMT by upregulating Twist1 and HIF-1α. The ability of STS to induce the Wnt/ß-catenin signaling and EMT has profound implications on estrogen-mediated carcinogenesis in human cancer cells.

13.
Biomol Ther (Seoul) ; 25(3): 321-328, 2017 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-27956712

RESUMEN

Steroid sulfatase (STS) is an enzyme responsible for the hydrolysis of aryl and alkyl sulfates. STS plays a pivotal role in the regulation of estrogens and androgens that promote the growth of hormone-dependent tumors, such as those of breast or prostate cancer. However, the molecular function of STS in tumor growth is still not clear. To elucidate the role of STS in cancer cell proliferation, we investigated whether STS is able to regulate the integrin signaling pathway. We found that overexpression of STS in HeLa cells increases the protein and mRNA levels of integrin ß1 and fibronectin, a ligand of integrin α5ß1. Dehydroepiandrosterone (DHEA), one of the main metabolites of STS, also increases mRNA and protein expression of integrin ß1 and fibronectin. Further, STS expression and DHEA treatment enhanced phosphorylation of focal adhesion kinase (FAK) at the Tyr 925 residue. Moreover, increased phosphorylation of ERK at Thr 202 and Tyr 204 residues by STS indicates that STS activates the MAPK/ ERK pathway. In conclusion, these results suggest that STS expression and DHEA treatment may enhance MAPK/ERK signaling through up-regulation of integrin ß1 and activation of FAK.

14.
J Cancer ; 7(6): 681-6, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27076849

RESUMEN

PURPOSE: The colorectal cancer (CRC) is the third leading cause of death in Korea. Ulcerative colitis (UC) is regarded as a risk factor of CRC. The aim of study is to confirm the incidence of CRC among subjects with and without a diagnosis of UC based on a sample of the Korean population. This study identified the effect of UC on incidence of CRC in Korea. METHOD: The data were from the population-based cohort containing National Health Insurance (NHI) claims from 2002 to 2013. We washed out first year (2002) for newly detected cases. Subjects who were under 20 years of age, diagnosed UC and CRC in 2002 development of CRC before diagnosis of UC since 2003, were excluded from analyses. Among 745,641 subjects during 11 years of follow-up (2003-2013), 7,448 patients with CRC were newly detected. Cox proportional hazard regression model was used to estimate the hazard ratio (HR) of UC for CRC incidence. Confounding variables including gender, baseline age, type of social security, income level, residence, Charlson Comorbidity Index, hypertension and diabetes mellitus were incorporated into the model. RESULTS: Overall annual incidence of UC and CRC were 6.7 and 95.4 per 100,000 during 11 years (2003~2013), respectively. Among 522 of newly detected UC cases, CRC incident cases were 12 cases during 11 years. The effects were stronger for male. Advancing age and Charlson Comorbidity Index, hypertension and diabetes mellitus increased the risk of CRC. This study showed that the adjusted hazard ratio of UC in incidence of CRC is 1.92 (95% confidence interval: 1.09-3.38). Also, male patients with UC have more HR than female patients with UC. CONCLUSION: The results of this study showed that patients with UC are the high risk group in incidence of CRC. Furthermore, the effects of UC in male patients are higher than those in female. The future study is needed to identify the effect of UC on mortality of CRC.

15.
PLoS One ; 11(3): e0151598, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26981862

RESUMEN

Cytochrome P450 1B1 (CYP1B1) is a major E2 hydroxylase involved in the metabolism of potential carcinogens. CYP1B1 expression has been reported to be higher in tumors compared to normal tissues, especially in hormone-related cancers including breast, ovary, and prostate tumors. To explore the role of CYP1B1 in cancer progression, we investigated the action of CYP1B1 in cells with increased CYP1B1 via the inducer 7,12-dimethylbenz[α]anthracene (DMBA) or an overexpression vector, in addition to decreased CYP1B1 via the inhibitor tetramethoxystilbene (TMS) or siRNA knockdown. We observed that CYP1B1 promoted cell proliferation, migration, and invasion in MCF-7 and MCF-10A cells. To understand its molecular mechanism, we measured key oncogenic proteins including ß-catenin, c-Myc, ZEB2, and matrix metalloproteinases following CYP1B1 modulation. CYP1B1 induced epithelial-mesenchymal transition (EMT) and activated Wnt/ß-catenin signaling via upregulation of CTNNB1, ZEB2, SNAI1, and TWIST1. Sp1, a transcription factor involved in cell growth and metastasis, was positively regulated by CYP1B1, and suppression of Sp1 expression by siRNA or DNA binding activity using mithramycin A blocked oncogenic transformation by CYP1B1. Therefore, we suggest that Sp1 acts as a key mediator for CYP1B1 action. Treatment with 4-hydroxyestradiol (4-OHE2), a major metabolite generated by CYP1B1, showed similar effects as CYP1B1 overexpression, indicating that CYP1B1 activity mediated various oncogenic events in cells. In conclusion, our data suggests that CYP1B1 promotes cell proliferation and metastasis by inducing EMT and Wnt/ß-catenin signaling via Sp1 induction.


Asunto(s)
Proliferación Celular/efectos de los fármacos , Citocromo P-450 CYP1B1/fisiología , Transición Epitelial-Mesenquimal , Metástasis de la Neoplasia , Transducción de Señal , Factor de Transcripción Sp1/metabolismo , Proteínas Wnt/metabolismo , beta Catenina/metabolismo , Línea Celular Tumoral , Citocromo P-450 CYP1B1/antagonistas & inhibidores , Citocromo P-450 CYP1B1/biosíntesis , Citocromo P-450 CYP1B1/genética , Estrógenos de Catecol/metabolismo , Humanos , Regulación hacia Arriba
16.
Toxicol Lett ; 223(2): 109-15, 2013 Nov 25.
Artículo en Inglés | MEDLINE | ID: mdl-24055520

RESUMEN

Human steroid sulfatase (STS) plays an important role in regulating the formation of biologically active estrogens and may be a promising target for treating estrogen-mediated carcinogenesis. The molecular mechanism of STS gene expression, however, is still not clear. Growth factors are known to increase STS activity but the changes in STS expression have not been completely understood. To determine whether insulin-like growth factor (IGF)-II can induce STS gene expression, the effects of IGF-II on STS expression were studied in PC-3 human prostate cancer cells. RT-PCR and Western blot analysis showed that IGF-II treatment significantly increased the expression of STS mRNA and protein in concentration- and time-dependent manners. To understand the signaling pathway by which IGF-II induces STS gene expression, the effects of specific PI3-kinase/Akt and NF-κB inhibitors were determined. When the cells were treated with IGF-II and PI3-kinase/Akt inhibitors, such as LY294002, wortmannin, or Akt inhibitor IV, STS expression induced by IGF-II was significantly blocked. Moreover, we found that NF-κB inhibitors, such as MG-132, bortezomib, Bay 11-7082 or Nemo binding domain (NBD) binding peptide, also strongly prevented IGF-II from inducing STS gene expression. We assessed whether IGF-II activates STS promoter activity using transient transfection with a luciferase reporter. IGF-II significantly stimulated STS reporter activity. Furthermore, IGF-II induced expression of 17ß-hydroxysteroid dehydrogenase (HSD) 1 and 3, whereas it reduced estrone sulfotransferase (EST) gene expression, causing enhanced estrone and ß-estradiol production. Taken together, these results strongly suggest that IGF-II induces STS expression via a PI3-kinase/Akt-NF-κB signaling pathway in PC-3 cells and may induce estrogen production and estrogen-mediated carcinogenesis.


Asunto(s)
Regulación Enzimológica de la Expresión Génica , Factor II del Crecimiento Similar a la Insulina/farmacología , Esteril-Sulfatasa/metabolismo , 17-Hidroxiesteroide Deshidrogenasas/genética , 17-Hidroxiesteroide Deshidrogenasas/metabolismo , Androstadienos/farmacología , Ácidos Borónicos/farmacología , Bortezomib , Línea Celular Tumoral , Cromonas/farmacología , Inhibidores Enzimáticos/farmacología , Humanos , Leupeptinas/farmacología , Masculino , Morfolinas/farmacología , FN-kappa B/antagonistas & inhibidores , FN-kappa B/genética , FN-kappa B/metabolismo , Nitrilos/farmacología , Fosfatidilinositol 3-Quinasas/genética , Fosfatidilinositol 3-Quinasas/metabolismo , Inhibidores de las Quinasa Fosfoinosítidos-3 , Reacción en Cadena de la Polimerasa , Neoplasias de la Próstata , Proteínas Proto-Oncogénicas c-akt/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Pirazinas/farmacología , Análisis de Secuencia de ADN , Transducción de Señal , Esteril-Sulfatasa/genética , Sulfonas/farmacología , Sulfotransferasas/antagonistas & inhibidores , Sulfotransferasas/genética , Sulfotransferasas/metabolismo , Wortmanina
17.
Biomol Ther (Seoul) ; 20(6): 556-61, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-24009850

RESUMEN

Steroid sulfatase (STS) is responsiblefor the conversion of estrone sulfate to estrone that can stimulate growth in endocrine-dependent tumors such as prostate cancer. Although STS is considered as a therapeutic target for the estrogen-dependent diseases, cellular function of STS are still not clear. Previously, we found that tumor necrosis factor (TNF)-α significantly enhances steroid sulfatase expression in PC-3 human prostate cancer cells through PI3K/Akt-dependent pathways. Here, we studied whether bacterial lipopolysaccharides (LPS) which are known to induce TNF-α may increase STS expression. Treatment with LPS in PC-3 cells induced STS mRNA and protein in concentration- and time-dependent manners. Using luciferase reporter assay, we found that LPS enhanced STS promoter activity. Moreover, STS expression induced by LPS increased PC-3 tumor cell migration determined by wound healing assay. We investigated that LPS induced IL-6 expression and IL-6 increased STS expression. Taken together, these data strongly suggest that LPS induces STS expression through IL-6 pathway in human prostate cancer cells.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA